3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      In vivo base editing by a single i.v. vector injection for treatment of hemoglobinopathies

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Individuals with β-thalassemia or sickle cell disease and hereditary persistence of fetal hemoglobin (HPFH) possessing 30% fetal hemoglobin (HbF) appear to be symptom free. Here, we used a nonintegrating HDAd5/35++ vector expressing a highly efficient and accurate version of an adenine base editor (ABE8e) to install, in vivo , a –113 A>G HPFH mutation in the γ-globin promoters in healthy CD46/β-YAC mice carrying the human β-globin locus. Our in vivo hematopoietic stem cell (HSC) editing/selection strategy involves only s.c. and i.v. injections and does not require myeloablation and HSC transplantation. In vivo HSC base editing in CD46/β-YAC mice resulted in > 60% –113 A>G conversion, with 30% γ-globin of β-globin expressed in 70% of erythrocytes. Importantly, no off-target editing at sites predicted by CIRCLE-Seq or in silico was detected. Furthermore, no critical alterations in the transcriptome of in vivo edited mice were found by RNA-Seq. In vitro, in HSCs from β-thalassemia and patients with sickle cell disease, transduction with the base editor vector mediated efficient –113 A>G conversion and reactivation of γ-globin expression with subsequent phenotypic correction of erythroid cells. Because our in vivo base editing strategy is safe and technically simple, it has the potential for clinical application in developing countries where hemoglobinopathies are prevalent.

          Related collections

          Most cited references56

          • Record: found
          • Abstract: found
          • Article: not found

          Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements

          CRISPR-Cas9 is poised to become the gene editing tool of choice in clinical contexts. Thus far, exploration of Cas9-induced genetic alterations has been limited to the immediate vicinity of the target site and distal off-target sequences, leading to the conclusion that CRISPR-Cas9 was reasonably specific. Here we report significant on-target mutagenesis, such as large deletions and more complex genomic rearrangements at the targeted sites in mouse embryonic stem cells, mouse hematopoietic progenitors and a human differentiated cell line. Using long-read sequencing and long-range PCR genotyping, we show that DNA breaks introduced by single-guide RNA/Cas9 frequently resolved into deletions extending over many kilobases. Furthermore, lesions distal to the cut site and crossover events were identified. The observed genomic damage in mitotically active cells caused by CRISPR-Cas9 editing may have pathogenic consequences.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            CRISPR–Cas9 genome editing induces a p53-mediated DNA damage response

            Here, we report that genome editing by CRISPR-Cas9 induces a p53-mediated DNA damage response and cell cycle arrest in immortalized human retinal pigment epithelial cells, leading to a selection against cells with a functional p53 pathway. Inhibition of p53 prevents the damage response and increases the rate of homologous recombination from a donor template. These results suggest that p53 inhibition may improve the efficiency of genome editing of untransformed cells and that p53 function should be monitored when developing cell-based therapies utilizing CRISPR-Cas9.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Phage-assisted evolution of an adenine base editor with improved Cas domain compatibility and activity

              Applications of adenine base editors (ABEs) have been constrained by the limited compatibility of the deoxyadenosine deaminase component with Cas homologs other than SpCas9. We evolved the deaminase component of ABE7.10 using phage-assisted non-continuous and continuous evolution (PANCE and PACE), resulting in ABE8e. ABE8e contains eight additional mutations that increase activity (kapp) 590-fold compared with ABE7.10. ABE8e offers substantially improved editing efficiencies when paired with a variety of Cas9 or Cas12 homologs. ABE8e is more processive than ABE7.10, which could benefit screening, disrupting regulatory regions and multiplex base editing applications. A modest increase in Cas9-dependent and -independent DNA off-target editing, and in transcriptome-wide RNA off-target editing can be ameliorated by introducing additional mutations in the TadA-8e domain. Finally, we show that ABE8e can efficiently edit natural mutations in a GATA1 binding site in the BCL11A enhancer or the HBG promoter in human cells, targets which were poorly edited with ABE7.10. ABE8e broadens the effectiveness and applicability of adenine base editing.
                Bookmark

                Author and article information

                Contributors
                Journal
                JCI Insight
                JCI Insight
                JCI Insight
                JCI Insight
                American Society for Clinical Investigation
                2379-3708
                10 October 2022
                10 October 2022
                10 October 2022
                : 7
                : 19
                : e162939
                Affiliations
                [1 ]Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, Washington, USA.
                [2 ]Gene and Cell Therapy Center, Hematology Department, George Papanicolaou Hospital, Thessaloniki, Greece.
                [3 ]Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.
                [4 ]Department of Chemistry and Chemical Biology and
                [5 ]Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts, USA.
                [6 ]Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece.
                [7 ]School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece.
                [8 ]Hematological Laboratory, Second Department of Internal Medicine, Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece.
                [9 ]Stem and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
                [10 ]Department of Pathology, University of Washington, Seattle, Washington, USA.
                Author notes
                Address correspondence to: André Lieber, University of Washington, Box 357720, Seattle, Washington 98195, USA. Phone: 206.221.3973; Email: lieber00@ 123456uw.edu . Or to: Chang Li, University of Washington, Box 357720, Seattle, Washington 98195, USA. Phone: 206.543.0109; Email: cli1239@ 123456uw.edu .
                Author information
                http://orcid.org/0000-0003-1442-5168
                http://orcid.org/0000-0003-4421-971X
                http://orcid.org/0000-0002-4359-5417
                http://orcid.org/0000-0001-5949-4947
                http://orcid.org/0000-0002-9943-7557
                http://orcid.org/0000-0002-9849-1279
                Article
                162939
                10.1172/jci.insight.162939
                9675455
                36006707
                634122a1-b913-46be-a3d6-98048d1a6a69
                © 2022 Li et al.

                This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 22 June 2022
                : 19 August 2022
                Categories
                Research Article

                hematology,stem cells,gene therapy,hematopoietic stem cells,monogenic diseases

                Comments

                Comment on this article